WO2009115612A1 - Biomarqueurs de la polyarthrite rhumatoïde - Google Patents

Biomarqueurs de la polyarthrite rhumatoïde Download PDF

Info

Publication number
WO2009115612A1
WO2009115612A1 PCT/EP2009/053369 EP2009053369W WO2009115612A1 WO 2009115612 A1 WO2009115612 A1 WO 2009115612A1 EP 2009053369 W EP2009053369 W EP 2009053369W WO 2009115612 A1 WO2009115612 A1 WO 2009115612A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
rheumatoid arthritis
detection
polypeptide
Prior art date
Application number
PCT/EP2009/053369
Other languages
English (en)
Inventor
Veerle Somers
Klaartje Somers
Pieter Stinissen
Original Assignee
Universiteit Hasselt
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universiteit Hasselt filed Critical Universiteit Hasselt
Priority to EP09722261.6A priority Critical patent/EP2307451B1/fr
Priority to US12/737,281 priority patent/US9683031B2/en
Priority to ES09722261.6T priority patent/ES2584438T3/es
Publication of WO2009115612A1 publication Critical patent/WO2009115612A1/fr
Priority to US15/498,196 priority patent/US20180030122A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6854Immunoglobulins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/10Musculoskeletal or connective tissue disorders
    • G01N2800/101Diffuse connective tissue disease, e.g. Sjögren, Wegener's granulomatosis
    • G01N2800/102Arthritis; Rheumatoid arthritis, i.e. inflammation of peripheral joints

Definitions

  • the present invention relates to the diagnosis of autoimmune disorders, more specifically to the diagnosis of rheumatoid disorders, chronic autoimmune arthritis and even more specifically to the diagnosis of rheumatoid arthritis.
  • a biomarker panel is provided which can be used to detect if a subject has rheumatoid arthritis. Also described are methods of identification of such biomarkers.
  • RA Rheumatoid Arthritis
  • ACR American College of Rheumatology
  • RF rheumatoid factor
  • anti-CCP antibodies are predictive for RA development in undifferentiated arthritis patients, and presence of these markers in RA is associated with a worse disease course (1 1-15).
  • the sensitivity of anti-CCP antibodies is only moderate (67-68%) despite a high specificity of 95% (16;17). This would imply that 33% of RA patients is anti-CCP antibody negative (ACPA-).
  • ACPA- anti-CCP antibody negative
  • the reported sensitivity for ACPA testing in early disease is even lower (16).
  • recent findings indicate a potentially different aetiology and pathogenesis in ACPA negative and ACPA positive RA (18- 20). This heterogeneity implies the need for a panel of different markers to achieve an accurate diagnosis for the entire RA patient population. Summary
  • RA disease markers are crucial, particularly for early RA and RF negative (RF-) ACPA negative (ACPA-) RA.
  • RF- RF negative
  • ACPA- ACPA negative
  • the SAS procedure is based on phage display of a cDNA expression library, made from RA synovial tissue, which is screened with pooled serum from RA patients.
  • the present invention provides a set of biomarkers which are highly specific for RA patients, particularly RA patients with a disease duration of less than one year and serum which is RF negative and/or ACPA-negative.
  • methods for specifically detecting rheumatoid arthritis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-14 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14 and wherein ii) the presence of said antibody indicates that said mammal suffers from rheumatoid arthritis .
  • the mammal to be diagnosed is a human.
  • the body fluid derived from the mammal is blood plasma, blood serum or synovial fluid.
  • detection is via immune-enzymatic processes such as, but not limited to, enzyme-linked immunosorbant assays (ELISA), immunofluorescent techniques, radioimmunological assays (RIA) and immunoblotting and LINE blot.
  • ELISA enzyme-linked immunosorbant assays
  • RIA radioimmunological assays
  • detection is via flow cytometry.
  • the detection and/or quantification of at least one specific antibody according to the methods described herein may be indicative for rheumatoid arthritis.
  • the detection and/or quantification of at least one specific antibody is indicative for rheumatoid arthritis with a disease stage of less than 1 year.
  • the detection and/or quantification of at least one specific antibody is indicative for rheumatoid arthritis in a patient which body fluid is seronegative for rheumatoid factor and/or anti-CCP antibodies.
  • the methods described herein further comprise the detection of rheumatoid factor.
  • the methods involve the detection of antibodies against cyclic citrullinated peptides.
  • the methods may comprise the detection of rheumatoid factor as well as of antibodies against cyclic citrullinated peptides.
  • polypeptides i.e. at least one polypeptide
  • Such polypeptide consists essentially of a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1- 14.
  • SEQ ID NO: 1-14 a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1- 14.
  • sequence may be part of a larger polypeptide; however, it should be noted that this term also implies that the defined sequence is the most important part of the molecule.
  • polypeptide consisting essentially of a defined sequence is necessarily less than double the length of the defined sequence.
  • polypeptide consists of a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • compositions comprising at least one polypeptide comprising a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • the composition consists essentially of at least one polypeptide.
  • compositions are provided comprising at least two different polypeptides comprising a sequence represented by any of SEQ ID NO: 1-14 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • the compositions consist essentially of the at least two different polypeptides.
  • the polypeptides can be used as biomarkers or to assist in diagnosis, e.g. of rheumatoid arthritis. Accordingly, in a further aspect, the polypeptides and compositions described herein are envisaged for use as a diagnostic. According to yet a further aspect, the (in vitro) use of the polypeptides or compositions described herein for detecting the presence and/or quantity of specific antibodies to the polypeptide or to at least one polypeptide of said composition in a body fluid of a mammal is provided. According to specific embodiments, the mammal of which the body fluid is tested is a human. According to other specific embodiments, the body fluid is blood plasma, blood serum or synovial fluid.
  • detection occurs via immune-enzymatic processes comprising enzyme-linked immunosorbant assays (ELISA), immunofluorescent techniques, radioimmunological assays (RIA), immunoblotting and LINE blot.
  • ELISA enzyme-linked immunosorbant assays
  • RIA radioimmunological assays
  • flow cytometry is used for detection.
  • the detection and/or quantification of at least one specific antibody is indicative for rheumatoid arthritis.
  • the detection and/or quantification of at least one specific antibody is indicative for rheumatoid arthritis with a disease stage of less than 1 year.
  • the detection and/or quantification of at least one specific antibody is indicative for rheumatoid arthritis in a patient which body fluid is seronegative for rheumatoid factor and/or anti-CCP antibodies.
  • the uses described herein further comprise the detection of rheumatoid factor.
  • the uses involve the detection of antibodies against cyclic citrullinated peptides.
  • the uses may comprise the detection of rheumatoid factor as well as of antibodies against cyclic citrullinated peptides.
  • methods for selecting a patient for a specific therapeutic treatment of rheumatoid arthritis or evaluating the therapeutic treatment of rheumatoid arthritis in a patient comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said patient wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-14 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14 and wherein ii) the decreased or increased concentration of said antibody leads to an election of a specific therapeutic treatment of rheumatoid arthritis in said patient.
  • diagnostic kits for the detection of rheumatoid arthritis comprising at least one polypeptide as described herein, or a composition as described herein, as well as reagents for making a medium appropriate for an immunological reaction to occur and reagents enabling to detect the antigen/antibody complex which has been produced by said immunological reaction.
  • antibodies are provided specifically binding to a polypeptide consisting essentially of a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • the antibodies do not bind to the full size proteins HLA-A, ribosomal protein S6 and MCM2.
  • the antibodies described herein are specific to polypeptides consisting essentially of a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14 and do not bind to the cognate full protein.
  • an antibody is provided against SEQ ID NO: 1.
  • FIG. 1 Serological Antigen Selection procedure.
  • the selection procedure entails incubation of phage displaying the RA cDNA library with pooled RA serum (1 ), leading to formation of phage antigen-lgG complexes (2). These complexes are captured on a solid support by anti- human IgG antibody (3), and non-bound phage are washed away (4). Bound phage are eluted and amplified through infection of host bacteria (5). Phage are produced again and used as input in a subsequent selection round (6).
  • Figure 2 Antibody levels against UH-RA.21 clone (a) and UH-RA.10 clone (b) in patients with RA, other rheumatic diseases and healthy controls (HC).
  • Scatterplots show mean optical density (OD)(RA-clone):OD(empty phage) ratios of 48 RA patients, 30 patients with other inflammatory rheumatic diseases and 38 healthy controls. Each symbol represents the mean of duplicate or triplicate measurements for each serum sample. The horizontal line constitutes the cut off value for a positive signal, i.e. 3 standard deviation (SD) above the mean OD(phage):OD(empty phage) ratio of the HC group.
  • SD standard deviation
  • Figure 4 Comparison of antibody levels against identified RA candidate antigens between patients with RA, control patients with other rheumatic diseases and healthy controls (HC). Scatterplots show mean OD(RA-clone)/OD(empty phage) ratios of 48 RA patients, 30 patients with other inflammatory rheumatic diseases and 38 healthy controls.
  • A UH-RA.9
  • B UH- RA.10
  • C UH-RA.13
  • D UH-RA.14
  • E UH-RA.17
  • F UH-RA.21
  • G UH-RA.22.
  • Antibody levels against UH-RA.21 and UH-RA.10 were significantly higher in RA patients compared to patients with other rheumatic diseases and healthy controls (P ⁇ 0.001 ).
  • Antibody levels against UH-RA.9, UH-RA.13, UH-RA.14, UH-RA.17 and UH-RA.22 were significantly higher in patients with RA compared to the HC group (PO.001 , PO.01 , PO.001 , PO.001 and PO.001 respectively).
  • Figure 5 Comparison of antibody levels against identified RA candidate antigens between patients with RA, control patients with other rheumatic diseases and healthy controls (HC). Scatterplots show mean OD(RA-clone)/OD(empty phage) ratios of 48 RA patients, 30 patients with other inflammatory rheumatic diseases and 38 healthy controls.
  • A UH-RA.1
  • B UH- RA.2,
  • C UH-RA.7
  • D UH-RA.1 1
  • E UH-RA.15
  • F UH-RA.16
  • G UH-RA.20. No significant differences in antibody levels between RA patients, patients with other rheumatic diseases and healthy controls could be discerned.
  • Each symbol represents the mean of triplicate measurements for each serum sample on tested phage and empty phage.
  • the horizontal line constitutes the cut-off value for a positive signal, i.e. 3SD above the mean OD(phage)/OD(empty phage) ratio of the HC group.
  • Figure 6 Specificity of RA immunoreactivity towards displayed cDNA products tested by peptide and competition ELISA.
  • A Representative example of reactivity against coated synthetic UH-RA.21 peptide (white bars) in 10 RA samples (RA-p.1 to RA-p.10). As a negative control, serum samples were also incubated with a non-relevant synthetic peptide (black bars).
  • B Competition of phage displaying the corresponding UH-RA.21 peptide with coated peptide demonstrated specificity of the serum antibodies against the phage displayed peptide.
  • UH-RA.21 phage particles were pre-incubated with anti-UH-RA.21 antibody-positive RA serum (black full line) (determined by phage ELISA), followed by transfer of this mix to coated UH-RA.21 peptide in a synthetic peptide ELISA format.
  • an antibody-negative RA serum (dotted line) was pre-incubated with UH-RA.21 displaying phage and was treated in an identical manner as the antibody-positive serum.
  • C As a negative control, antibody-positive (black full line) and antibody-negative (dotted line) RA serum samples were pre-incubated with increasing amounts of empty phage particles.
  • Figure 7 Results of a synthetic peptide ELISA screening on serum samples from RA patients and different control groups for anti-UH-RA.21 immunoreactivity. Eighteen out of 78 RA patients (23%), 6 out of 49 OA patients (12%), 5 out of 26 psoriatic arthritis patients (19%), 5 out of 35 ankylosing spondylitis patients (14%), 2 out of 48 HC (4%) and 1 out of 42 subjects with mechanical joint complaints (2%) were antibody-positive for UH-RA.21. Significant differences in antibody-reactivity towards UH-RA.21 were demonstrated between RA patients and the combined group of HC and mechanical joint complaints subjects on the one hand, and between RA patients and OA patients on the other hand. The ratios of OD 450 measured for UH- RA.21 to OD 450 measured for the non-relevant peptide are shown for all tested patients.
  • Arthritic diseases include both seropositive rheumatoid arthritis (RA) and seronegative chronic arthritis, a prototypic group of autoimmune diseases characterised by non-reactivity of serum antibodies against rheumatoid factor (RF) or citrullinated protein antigens (ACPA). These subtypes include seronegative rheumatoid arthritis (RA), (30% of RA patients remain negative for either RF or ACPA), spondyloarthropathy (SpA), psoriatic arthritis (Psa) and juvenile idiopathic arthritis (JIA). These diseases have a relatively high prevalence (ranging from 1 in 100 for RA), cannot be cured and are associated with high morbidity.
  • RA seropositive rheumatoid arthritis
  • RF rheumatoid factor
  • ACPA citrullinated protein antigens
  • Biomarker panel which can be used for the detection of chronic autoimmune arthritis, more specifically for the detection of rheumatoid arthritis (RA) in patients.
  • Biomarkers were isolated with the technology of Serological Antigen Selection (SAS) wherein antigens (i.e. biomarkers) were identified that bind to antibodies present in serum of patients suffering from rheumatoid arthritis.
  • SAS Serological Antigen Selection
  • a cDNA phage display library comprising cDNA products derived from RA synovial tissue - expressed as a fusion to minor coat protein pVI of filamentous phage M13 - was panned to identify cDNA clones that bind auto-antibodies in serum specimens from RA patients.
  • the RA cDNA library was selected on two pools, the first consisting of sera from RA patients with disease duration of less than one year, the second containing sera from RF negative and ACPA negative RA patients.
  • a biomarker panel of 14 antigenic cDNA targets with high specificity for RA was retrieved.
  • the invention provides polypeptides (i.e. at least one polypeptide) that can be used as biomarkers or to assist in diagnosis, e.g. of rheumatoid arthritis.
  • polypeptide consists essentially of a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • compositions are also provided comprising at least one polypeptide comprising a sequence selected from SEQ ID NO: 1-14 or a fragment thereof comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • the composition consist essentially of the at least one polypeptide.
  • compositions comprising at least two different polypeptides comprising a sequence represented by any of SEQ ID NO: 1-14 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • These polypeptides or compositions are herein also designated as a biomarker or as a biomarker panel or as a set of biomarkers.
  • the SEQ ID NO: 1-14 correspond with the translated amino acid sequences of the antigens retrieved by the selection of phage displayed RA cDNA expression library on serum fluid derived from RA patients.
  • the translation of the insert of UH-RA.21 corresponds with SEQ ID NO: 1 , that of UH-RA.1 1 with SEQ ID NO: 2, of UH-RA.14 with SEQ ID NO: 3, of UH-RA.15 with SEQ ID NO: 4, of UH-RA.16 with SEQ ID NO: 5, of UH-RA.1 with SEQ ID NO: 6, of UH- RA.2 with SEQ ID NO: 7, of UH-RA.7 with SEQ ID NO: 8, of UH-RA.9 with SEQ ID NO: 9, of UH-RA.10 with SEQ ID NO: 10, UH-RA.13 with SEQ ID NO: 1 1 , of UH-RA.17 with SEQ ID NO: 12, of UH-RA.20 with SEQ ID NO: 13, and the translation of the insert of UH-RA.22 corresponds with SEQ ID NO: 14 (see table 7).
  • SEQ ID NO: 15-28 The nucleotide sequences which encode SEQ ID NO: 1-14 are depicted in SEQ ID NO: 15-28 (wherein SEQ ID NO: 15 encodes SEQ ID NO: 1 , ..., and SEQ ID NO: 28 encodes SEQ ID NO: 14).
  • SEQ ID NO: 15-28 one or more nucleic acids of the invention may also be used as a biomarker, e.g. by having them transcribed or translated; or by detecting the nucleic acids directly instead of detecting the proteins, e.g. via quantitative PCR.
  • a composition comprises at least one polypeptide, or possibly at least two polypeptides, wherein such a polypeptide comprises a sequence as depicted by SEQ ID NO: 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13 or 14.
  • a polypeptide present in the composition can also be a protein, particularly when the composition contains at least two polypeptides.
  • SEQ ID NO: 12 was cloned as a partial 76 amino acid fragment of Selenoprotein P (protein product as detected using SAS). Since SEQ ID NO: 12 (corresponding with UH- RA.17) is a fragment of the selenoprotein P protein the composition can also comprise the full length selenoprotein P protein.
  • composition of the invention can also comprise at least one, or at least two different, polypeptide(s) wherein said polypeptide(s) are fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13 or 14. It is envisaged that 5 consecutive amino acids derived from SEQ ID NO: 1-14 are sufficient to be recognized as antigens by the auto-antibodies present in for example blood serum or blood plasma.
  • the fragments of the polypeptides comprise at least 6 consecutive amino acids derived from SEQ ID NO: 1-14, or at least 7, at least 8, at least 9, at least 10, at least 1 1 , at least 12, at least 13, at least 14, at least 15 or at least 20 amino acids derived from SEQ ID NO: 1-14.
  • the polypeptide sequences used are not completely identical to SEQ ID NO: 1-14, but almost identical. For instance, they show 99% sequence identity, 98% sequence identity, 95% sequence identity, 90% sequence identity, or 85% sequence identity to any of SEQ ID NO: 1-14. Percentage sequence identity is calculated according to methods known in the art, e.g. the BLAST algorithm. The following terms are typically used to describe the sequence relationships between two or more nucleic acids or polynucleotides: (a) "reference sequence", (b) "comparison window”, and (c) "percentage of sequence identity". (a) As used herein, "reference sequence” is a defined sequence used as a basis for sequence comparison. A reference sequence may be a subset or the entirety of a specified sequence; for example, SEQ ID NO:1 or a fragment of at least 5 consecutive amino acids thereof.
  • comparison window makes reference to a contiguous and specified segment of a polynucleotide or amino acid sequence, wherein the polynucleotide or amino acid sequence may be compared to a reference sequence and wherein the portion of the polynucleotide or amino acid sequence in the comparison window may comprise additions or deletions (i.e., gaps) compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • a gap penalty is typically introduced and is subtracted from the number of matches. Note that to align sequences dissimilar in length, the comparison window will usually be determined using the shorter of the two sequences.
  • percentage of sequence identity means the value determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide or amino acid sequence in the comparison window may comprise additions or deletions (i.e. gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.
  • the polypeptide sequences used are not completely identical to SEQ ID NO: 1-14, but highly similar, i.e. amino acids have been conservatively substituted. This implies that one or more hydrophobic amino acids have been replaced with other hydrophobic amino acids (or vice versa for hydrophilic amino acids), one or more positively charged amino acids have been replaced with other positively charged amino acids (or vice versa for negatively charged amino acids), and so on. Percentage similarity can be determined by the skilled person, e.g. using the BLAST program. In particular, the sequences used are 99% similar, 98% similar, 97% similar, 95% similar, 90% similar or 85% similar to any one of SEQ ID NO: 1-14.
  • composition comprises 14 different polypeptides comprising a sequence selected from SEQ ID NO: 1-14 or 14 different fragments comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14.
  • the invention provides the use of a composition of the invention for detecting the presence of specific antibodies to at least one polypeptide present in said composition wherein said antibodies are present in a body fluid of a mammal.
  • the invention provides the use of a composition of the invention for detecting the presence of specific auto-antibodies to at least one polypeptide present in said composition wherein said auto-antibodies are present in a body fluid of a mammal.
  • said use of a composition is an "in vitro" use of a composition. The latter implies a diagnostic method with no direct interaction with the patient.
  • the invention provides the use of a composition of the invention for the manufacture of a diagnostic assay to detect autoimmune disorders, more specifically to the diagnosis of rheumatoid disorders, chronic autoimmune arthritis and even more specifically to the diagnosis of rheumatoid arthritis.
  • body fluid includes blood, blood serum, blood plasma, saliva, urine, tears, bone marrow fluid, cerebrospinal fluid (CSF), synovial fluid, lymphatic fluid, amniotic fluid, nipple aspiration fluid and the like.
  • Preferred body fluids for analysis are those that are conveniently obtained from patients, particularly preferred body fluids include blood serum, blood plasma and synovial fluid.
  • the invention provides a method for detecting rheumatoid arthritis in a mammal comprising i) detecting the presence of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-14 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14 and wherein ii) the presence of said antibody indicates that said mammal suffers from rheumatoid arthritis .
  • the invention provides a method for detecting rheumatoid arthritis with a disease stage of less than one year.
  • the invention provides a method for detecting rheumatoid arthritis in a body fluid derived from a mammal which is seronegative for rheumatoid factor and/or anti- CCP antibodies.
  • the method for detecting rheumatoid arthritis in a mammal of the present invention is combined with the detection of rheumatoid factor, citrullinated peptides (e.g.
  • the invention provides a method for evaluating the prognosis/disease severity of rheumatoid arthritis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1-14 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14 and wherein ii) the presence (decreased or increased) concentration of said antibody indicates the prognosis of rheumatoid arthritis in said mammal.
  • the invention provides a method for selecting mammals for a specific therapeutic treatment of rheumatoid arthritis or evaluating the therapeutic treatment of rheumatoid arthritis in a mammal comprising i) detecting the presence or quantity of at least one antibody in a body fluid derived from said mammal wherein said antibody has a specificity for a polypeptide comprising a sequence selected from the group consisting of SEQ ID NO: 1- 14 or a fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1- 14 and wherein ii) the presence or increased concentration of said antibody leads to an election of a specific therapeutic treatment of rheumatoid arthritis in said mammal.
  • said body fluid is serum, plasma or synovial fluid.
  • said mammal is a human.
  • the invention provides an antibody that specifically binds to a polypeptide selected from the group consisting of polypeptides represented by SEQ ID NO: 1- 14.
  • a polypeptide selected from the group consisting of polypeptides represented by SEQ ID NO: 1- 14.
  • Methods for generating antibodies are well known in the art.
  • the antibodies are monoclonal antibodies.
  • the polypeptides forming part of the compositions of the invention may be synthesized chemically or may be made in a recombinant way. They may also be coupled to a soluble carrier after synthesis or after recombinant production. If a carrier is used the nature of such a carrier should be such that it has a molecular weight greater than 5000 and should not be recognized by antibodies.
  • a carrier can be a protein.
  • Proteins which are frequently used as carriers are keyhole limpet hemocyanin, bovine gamma globulin, bovine serum albumin, and poly-L- lysine. There are many well described techniques for coupling peptides to carriers. The linkage may occur at the N-terminus, C-terminus or at an internal site in the peptide.
  • the polypeptide may also be derivatized for coupling.
  • the polypeptides may also be synthesized directly on an oligo-lysine core in which both the alpha as well as the epsilon-amino groups of lysines are used as growth points for the polypeptides.
  • the number of lysines comprising the core is preferably 3 or 7. Additionally, a cysteine may be included near or at the C-terminus of the complex to facilitate the formation of homo- or heterodimers.
  • the invention relates to a process for detecting antibodies (i.e. auto- antibodies) related to RA or other rheumatic/autoimmune disorders in a biological sample (such as serum or plasma) of a mammal liable to contain them, this process comprising contacting the biological sample with a composition according to the invention under conditions enabling an immunological reaction between said composition and the antibodies which are possibly present in the biological sample and the detection of the antigen/antibody complex which may be formed.
  • the detection can be carried out according to any classical process.
  • immune-enzymatic processes according to the ELISA technique or immunofluorescent or radioimmunological (RIA) or the equivalent ones can be used.
  • the invention also relates to polypeptides according to the invention labeled by an appropriate label of the enzymatic, fluorescent, biotin, radioactive type.
  • a method for detecting antibodies related to RA comprises for instance the following steps: deposit of determined amounts of a polypeptidic composition according to the invention on a support (e.g. into wells of a titration microplate), introduction on said support (e.g. into wells) of increasing dilutions of the body fluid (e.g. serum) to be diagnosed, incubation of the support (e.g. microplate), repeated rinsing of the support (e.g.
  • microplate introduction on the support labeled antibodies which are specific for immunoglobulins present in the body fluid, the labeling of these antibodies being based on the activity of an enzyme which is selected from among the ones which are able to hydrolyze a substrate by modifying the absorption of the radiation of this latter at least at a given wave length, detection by comparing with a control standard of the amount of hydrolyzed substrate.
  • the invention also relates to a process for detecting and identifying antigens of RA in a body specimen (tissue, biopsy, ...) liable to contain them, this process comprising: contacting the biological sample with an appropriate antibody of the invention (i.e. antibodies with a specificity for a polypeptide of the composition) under conditions enabling an immunological reaction between said antibody and the antigens of RA which are possibly present in the biological sample and the detection of the antigen/antibody complex which may be formed.
  • an appropriate antibody of the invention i.e. antibodies with a specificity for a polypeptide of the composition
  • antibodies in particular auto-antibodies, which recognize the polypeptides of the invention, can be detected in a variety of ways.
  • One method of detection is further described in the examples and uses enzyme-linked immunosorbant assay (ELISA) of the polypeptides of the invention displayed by phages (i.e. phage-ELISA technology).
  • ELISA enzyme-linked immunosorbant assay
  • this will be a microtiter plate but may in principle be any sort of insoluble solid phase (e.g. glass, nitrocellulose).
  • a suitable dilution or dilutions of for example serum to be tested is brought into contact with the solid phase to which the polypeptide is bound.
  • a solution hybridization is carried out in which high affinity interactions occur (eg. biotinylated polypeptides of the composition are pre-incubated with serum). The incubation is carried out for a time necessary to allow the binding reaction to occur. Subsequently, unbound components are removed by washing the solid phase. The detection of immune complexes (i.e.
  • auto-antibodies present in for example human serum binding to at least one polypeptide of the invention is achieved using antibodies which specifically bind to human immunoglobulins, and which have been labeled with an enzyme, preferably but not limited to either horseradish peroxidase, alkaline phosphatase, or beta-galactosidase, which is capable of converting a colorless or nearly colorless substrate or co-substrate into a highly colored product or a product capable of forming a colored complex with a chromogen.
  • an enzyme preferably but not limited to either horseradish peroxidase, alkaline phosphatase, or beta-galactosidase, which is capable of converting a colorless or nearly colorless substrate or co-substrate into a highly colored product or a product capable of forming a colored complex with a chromogen.
  • the detection system may employ an enzyme which, in the presence of the proper substrate(s), emits light.
  • the amount of product formed is detected either visually, spectrophotometrically, electrochemically, fluorescently or luminometrically, and is compared to a similarly treated control.
  • the detection system may also employ radioactively labeled antibodies, in which case the amount of immune complex is quantified by scintillation counting or gamma counting.
  • Other detection systems which may be used include those based on the use of protein A derived from Staphylococcus aureus Cowan strain I, protein G from group C Staphylococcus sp. (strain 26RP66), or systems which make use of the high affinity biotin-avidin or streptavidin binding reaction.
  • polypeptides of the invention may be either labeled or unlabeled. Labels which may be employed may be of any type, such as enzymatic, chemical, fluorescent, luminescent, or radioactive.
  • the polypeptides may be modified for binding to surfaces or solid phases, such as, for example, microtiter plates, nylon membranes, glass or plastic beads, and chromatographic supports such as cellulose, silica, or agarose. The methods by which polypeptides can be attached or bound to solid support or surface are well known to those skilled in the art.
  • the polypeptides of the invention can be prepared according to the classical techniques in the field of peptide synthesis.
  • the synthesis can be carried out in homogeneous solution or in solid phase.
  • the synthesis technique in homogeneous solution which can be used is the one described by Houbenweyl in the book titled “Methode der organischen chemie” (Method of organic chemistry) edited by E. Wunsh, vol. 15-1 et II. THIEME, Stuttgart 1974.
  • the polypeptides of the invention can also be prepared in solid phase according to the method described by Atherton & Shepard in their book titled “Solid phase peptide synthesis” (Ed. IRL Press, Oxford, NY, Tokyo, 1989).
  • Synthesis protocols in the art generally employ the use of t- butyloxycarbonyl- or ⁇ -fluorenylmethoxy-carbonyl-protected activated amino acids.
  • the procedures for carrying out the syntheses, the types of side-chain protection, and the cleavage methods are amply described in, for example, Stewart and Young, Solid Phase Peptide Synthesis, 2nd Edition, Pierce Chemical Company, 1984; and Atherton and Sheppard, Solid Phase Peptide Synthesis, IRL Press, 1989.
  • antibodies raised to polypeptides of the invention can also be used in conjunction with labeled polypeptides of the invention for the detection of (auto)-antibodies present in for example serum by competition assay.
  • antibodies raised to polypeptides are attached to a solid support which may be, for example, a plastic bead or a plastic tube. Labeled polypeptide is then mixed with suitable dilutions of the fluid (e.g. serum) to be tested and this mixture is subsequently brought into contact with the antibody bound to the solid support. After a suitable incubation period, the solid support is washed and the amount of labeled polypeptide is quantified.
  • a reduction in the amount of label bound to the solid support is indicative of the presence of (auto)-antibodies in the original sample.
  • the polypeptide may also be bound to the solid support. Labeled antibody may then be allowed to compete with (auto)-antibody present in the sample (e.g. serum) under conditions in which the amount of polypeptide is limiting. As in the previous example, a reduction in the measured signal is indicative of the presence of (auto)-antibodies in the sample tested.
  • a test for giving evidence of the fact that one or more polypeptides present in a composition of the invention are recognized by antibodies present in for example serum is an immunoblotting (or Western blotting) analysis or LINE assay.
  • polypeptides can be chemically synthesized or polypeptides (or the protein) can be produced via recombinant techniques.
  • polypeptides of the invention are blotted onto nitrocellulose membranes (e.g. Hybond C. (Amersham)) as described by Towbin H.
  • nitrocellulose sheets are incubated overnight with each of these samples (e.g. diluted 1 :50) (after blocking a-specific protein- binding sites). Reactive areas on the nitrocellulose sheets are revealed by incubation with e.g. peroxidase conjugated goat anti-human immunoglobulin G antibody (e.g. diluted 1 :200) for 4 h, and after repeated washings, color reaction is developed by adding for example alpha- chloronaphtol (Bio-Rad Laboratories, Richmond, Calif.) in the presence of hydrogen peroxide.
  • peroxidase conjugated goat anti-human immunoglobulin G antibody e.g. diluted 1 :200
  • the free reactive functions which are present in some of the amino acids, which are part of the constitution of the polypeptides of the invention particularly the free carboxyl groups which are carried by the groups GIu and Asp or by the C-terminal amino acid on the one hand and/or the free NH2 groups carried by the N-terminal amino acid or by amino acids inside the peptidic chain, for instance Lys, on the other hand, can be modified in so far as this modification does not alter the above mentioned properties of the polypeptide.
  • the polypeptides which are thus modified are naturally part of the invention.
  • the above mentioned carboxyl groups can be acylated or esterified. Other modifications are also part of the invention.
  • the amine or carboxyl functions or both of terminal amino acids can be themselves involved in the bond with other amino acids.
  • the N-terminal amino acid can be linked to the C-terminal amino acid of another peptide comprising from 1 to several amino acids.
  • any peptidic sequences resulting from the modification by substitution and/or by addition and/or by deletion of one or several amino acids of the polypeptides according to the invention are part of the invention in so far as this modification does not alter the above mentioned properties of said polypeptides.
  • the polypeptides according to the invention can be glycosylated or not, particularly in some of their glycosylation sites of the type Asn-X-Ser or Asn-X-Thr, X representing any amino acid.
  • An advantageous recombinant polypeptide included in the composition of the invention is SEQ ID NO: 1 (UH-RA.21 ) since this polypeptide shows the highest frequency of antibody responses in serum of RA patients.
  • polypeptides are also possible depending on its intended use.
  • the polypeptide may be synthesized with an extra cysteine residue added. This extra cysteine residue is preferably added to the amino terminus and facilitates the coupling of the polypeptide to a carrier protein which is necessary to render the small polypeptide immunogenic.
  • the polypeptide is to be labeled for use in radioimmune assays, it may be advantageous to synthesize the protein with a tyrosine attached to either the amino or carboxyl terminus to facilitate iodination.
  • polypeptide possesses therefore the primary sequence of the polypeptide above-mentioned but with additional amino acids which do not appear in the primary sequence of the protein and whose sole function is to confer the desired chemical properties to the polypeptide.
  • the invention provides for a kit to diagnose RA.
  • the following necessary or kit can be used, said necessary or kit comprising: a composition (comprising at least one polypeptide selected from SEQ ID NO: 1- 14) according to the invention, or at least one fragment comprising at least 5 consecutive amino acids derived from SEQ ID NO: 1-14, reagents for making a medium appropriate for the immunological reaction to occur, reagents enabling to detect the antigen/antibody complex which has been produced by the immunological reaction, said reagents possibly having a label, or being liable to be recognized by a labeled reagent, more particularly in the case where the above mentioned polypeptide is not labeled.
  • RA synovial tissue used for library construction was analysed immunohistochemically prior to use. Severe and ongoing inflammation was shown by the presence of inflammatory infiltrates consisting primarily of macrophages and T-lymphocytes. B-lymphocytes were less prominent and primarily localised perivascularly. No germinal center structures were detected.
  • Primary library construction into the ⁇ -Uni-ZAP XR vector resulted in the generation of a RA ⁇ - Uni-ZAP XR library with a primary diversity of 4.5 x 10 6 recombinants.
  • RA-pSPVI-A/B/C display libraries containing respectively 4.6 x 10 6 , 8.5 x 10 6 and 5.3 x 10 6 cfu. Sequencing of the resulting RA-pSPVI libraries showed the presence of cDNA sequences encoding known synovial and cartilage components (collagen, osteonectin), candidate RA antigens (for example fibronectin), and unknown genes.
  • colony PCR and DNA fingerprinting techniques were performed on output phage from selection rounds 2, 3 and 4. Application of these techniques generates characteristic restriction profiles for each cDNA clone. The increasing presence of a particular restriction profile with increasing selection round number indicates enrichment of this particular cDNA clone throughout the selection procedure.
  • 250 phage clones obtained from selections on both serum pools were screened by colony PCR and fingerprinting analysis (table 1 ). While the output phage from the second round still showed a high degree of diversity, from the third round on however, specific fingerprinting profiles became more prominent, indicating enrichment of specific clones (data not shown).
  • Phage ELISA screening of 22 enriched candidate clones with sera used for affinity selections To confirm that enrichment of the 22 candidate clones had occurred based on interactions with RA patient antibodies, immunoreactivities against the phage clones were analysed in the individual RA sera that made up the pools by means of a phage ELISA technique. Phage displaying a specific cDNA product were incubated with individual serum specimens, and if individual serum contained antibodies directed against the phage displayed cDNA product, formation of phage-antibody complexes occurred, followed by detection of these complexes.
  • phage ELISA was performed on all 22 clones using 38 RA samples that were not used for affinity selections, 38 healthy control sera and 30 sera from patients with other rheumatic inflammatory diseases (table 1 ) to analyse the frequency of antibody responses. Results are shown in table 5.
  • Screening for antibody reactivity towards the identified antigenic panel is of added value to ACPA and RF serological testing.
  • Antibody levels against the 14 identified clones measured with phage ELISA screening in all RA and control serum samples are depicted in figure 4 (UH-RA.9, UH-RA.10, UH-RA.13, UH- RA.14, UH-RA.17, UH-RA.21 and UH-RA.22) and in figure 5 (UH-RA.1 , UH-RA.2, UH-RA.7, UH-RA.1 1 , UH-RA.15, UH-RA.16 and UH-RA.20).
  • HLA-A UH-RA.11
  • ribosomal protein S6 UH-RA.16
  • MCM2 UH-RA.15
  • monoclonal or polyclonal antibodies could be commercially obtained. Staining with these antibodies was performed on synovial knee tissue from three RA patients, all characterised by severe joint destruction. As a negative control, synovial knee tissue from a patient with destructive gonarthrosis was used. For all three proteins, staining was detected in the synovial tissues from all 3 RA patients, in comparison to the absence of detectable staining in the rheumatic control sample (not shown). This complies with elevated expression of these proteins in inflamed RA tissue. For HLA-A and ribosomal protein S6, staining seemed to be associated with inflammatory infiltrates, while this could not be observed for MCM2.
  • human anti-UH- RA.21 antibodies were immuno-affinity purified from a positive RA serum sample by the use of UH-RA.21 coated beads, lmmunohistochemical staining with the polyclonal anti-UH-RA.21 antiserum on the synovial tissue from 3 RA patients and 1 control rheumatic patient was performed, resulting in a positive staining in one RA synovial tissue. In this tissue, staining was demonstrated to be predominantly located at inflammatory infiltrates and in the hyperproliferating synovial lining (not shown).
  • Synovial tissue obtained at total hip replacement surgery of a 64-year old male patient with a 20-year history of seropositive RA, was used for RA library construction.
  • the synovial tissue was analysed immunohistochemically by staining for the presence of inflammatory cells such as macrophages (CD68+), T-cells (CD3+) and B-cells (CD20+).
  • inflammatory cells such as macrophages (CD68+), T-cells (CD3+) and B-cells (CD20+).
  • PoIy(A)+ RNA was directly isolated (polyATtract system 1000, Promega, Leiden, the Netherlands) from the synovial tissue without prior total RNA isolation as described by the manufacturer.
  • Oligo(dT) linker-primed cDNA was cloned into a ⁇ -Uni-ZAP XR vector system (Stratagene, La JoIIa, USA) followed by packaging of ⁇ phage. After mass excision of the pBluescript SK(-) phagemid from the Uni- ZAP XR vector, cDNA inserts were obtained by Xhol-Xbal (New England Biolabs, Ipswich, USA) restriction digestion.
  • cDNA inserts (sizes 500-2500 bp) were gel-purified (GFX gel band purification kit, GE Healthcare, Diegem, Belgium) and subsequently directionally cloned into our Xhol-Xbal digested pSPVI-A/B/C phagemid vectors, each encoding a different reading frame.
  • cDNA cloning occurs via C-terminal fusion to the pVI phage minor coat protein, resulting in display of the encoded cDNA products at the phage surface (21 ;22).
  • RA-pSPVI-A/B/C display libraries were obtained. Sequencing was performed on each phage display library.
  • Serum samples were obtained from 48 RA patients, who were diagnosed based on fulfilment of the ACR criteria. Patient characteristics are summarized in table 1.
  • RF serology Determination of RF serology was performed with the Serodia-RA Particle Agglutination test (Fujirebio Diagnostics, Goteberg, Sweden) according to the manufacturer's instructions. ACPA measurements were performed with a second-generation (anti-CCP2) ELISA test (Quanta Lite CCP IgG, INOVA Diagnostics, San Diego, USA), with a cut off value of 20 IU to define a positive test.
  • affinity selections two serum pools from 10 RA patients each were used. Characteristics of patients used for affinity selections are shown in table 2 and 3. The first pool consisted of sera from randomly selected RA patients with early disease course (symptoms of less than 1 year) (table 2).
  • the Serological Antigen Selection procedure was applied on the RA cDNA phage display library as described (24;25). Four subsequent affinity selection rounds were performed in parallel with the early and RF- ACPA- RA serum pools. To enrich for phage clones based on high-affinity interactions with RA patient immunoglobulins (IgG), the selection procedure was performed more stringent with increasing rounds of selection.
  • IgG immunoglobulins
  • Sequence analysis Sequencing was performed on purified PCR products (GFX PCR DNA purification kit, GE Healthcare) of individual clones with primer 5'-TTA CCC TCT GAC TTT GTT CA-3' (Eurogentec), which annealed to the phagemid gene encoding pVI, and Big Dye TMT Terminator Cycle Sequence Ready Reaction Kit Il (Applied Biosystems, Warrington, United Kingdom), followed by analysis on an ABI Prism 310 Genetic Analyser (Applied Biosystems). Homology search analysis of nucleotide and amino acid sequences was performed with the basic local alignment search tool software of NCBI.
  • the level of antibody reactivity of individual serum samples against individual selected phage clones was measured by phage ELISA.
  • ELISA of ligand displaying phage was performed as described previously (23;24). Background reactivity against phage particles was accounted for by measuring the ELISA signal obtained by incubating the tested serum sample with empty phage, in parallel to incubation with the tested phage clone encoding a specific cDNA product.
  • a serum sample with an OD(tested phage):OD(empty phage) ratio higher than 3 times the standard deviation (SD) above the mean ratio OD(tested phage):OD(empty phage) of the HC group was considered positive for antibodies against the encoded cDNA product.
  • SD standard deviation
  • Fisher's exact test Associations between positivity for antibodies against particular cDNA clones and diagnosis of RA, were analysed by Fisher's exact test. Correlations between antibody levels and different clinical variables were examined by Spearman's correlation coefficient.
  • Recombinant protein expression and purification is performed according to the pBAD/TOPOThioFusion kit (Invitrogen, Merelbeke, Belgium).
  • the recombinant protein is expressed as a fusion protein to thioredoxin at the N-terminal side and a His6-tag at the C-term.
  • the cDNA insert encoding the candidate antigen was amplified and PCR products were cloned into the pBAD/TOPOThioFusion vector followed by transformation of the ligation mixtures into TOP10 E. coli bacteria.
  • plasmids were isolated and transformed into LMG194 E. coli bacteria.
  • LMG194 bacteria containing the pBAD/TOPO vector with insert, were cultured in LB medium (Invitrogen) to OD600 of 0.5 after which recombinant protein expression was induced during 4 hours by arabinose (0.2%) addition. After centrifugation, bacterial cells were resuspended in lysisbuffer (6M guanidiumchloride, 20 mM sodium phosphate, 500 mM NaCI, pH 7.8) followed by sonication for disruption of protein inclusion bodies. Recombinant protein purification was performed with Ni-NTA Sepharose beads (IBA, Leusden, the Netherlands) followed by dialysis against PBS.
  • lysisbuffer 6M guanidiumchloride, 20 mM sodium phosphate, 500 mM NaCI, pH 7.8
  • Protein identity was confirmed by performing SDS-PAGE followed by excision of the protein band and ESI-LC-MS/MS analysis (ThermoFinnigan, San Jose, US). The purified protein concentration was determined with the BCA protein quantification kit (Fisher Scientific, Doornik, Belgium). 9. Peptide/protein ELISA and competition ELISA
  • Synthetic purified peptides (Eurogentec) were coated at 1 ⁇ g/ml in PBS (pH 7.4) overnight at room temperature in peptide ELISA plates (polystyrene flat-bottom ELISA plates, Greiner Bio- One, Wemmel, Belgium). Recombinant proteins were coated at 1 ⁇ g/ml in carbonate buffer (0.1 M sodium hydrogen carbonate, pH 9.6) overnight at 4°C in ELISA plates (Greiner Bio- One). After washing with 0.05% PBS-Tween20 (PBS-T), blocking was performed with 2% milk powder in PBS (M-PBS) for 2 hours at 37°C.
  • a ratio of OD(tested peptide)/OD(non-relevant peptide) of more than 1 .5 was considered a positive signal.
  • a ratio of OD(protein)/OD(thioredoxin) of more than 1.5 was used as cut-off for a positive signal. Samples were tested in duplicate within a single experiment, and experiments were performed in duplicate. To further confirm specificity of measured antibodies towards phage displayed cDNA products, a competition ELISA between purified peptide and phage displaying the corresponding peptide was applied.
  • the serum samples were pre-incubated with increasing amounts of phage displaying the peptide at the surface. Competition of phage-displayed peptide with the coated peptide for serum antibodies results in decreased OD signals. As a negative control, increasing amounts of empty phage were pre-incubated with the serum samples.
  • Paraffin-embedded sections were mounted on polysine-coated glass slides (VWR, Heverlee, Belgium) and after dewaxing and rehydrating, endogenous peroxidase activity was blocked by incubation of the slides in a 0.3% hydrogen peroxide in methanol solution during 10 minutes.
  • endogenous peroxidase activity was blocked by incubation of the slides in a 0.3% hydrogen peroxide in methanol solution during 10 minutes.
  • prevention of aspecific binding was performed by incubation of the sections with protein block (Dako) during 20 minutes at room temperature.
  • the synovial tissue sections were blocked with non-conjugated secondary antibody, namely rabbit anti-human IgG polyclonal antibody (Dako) diluted 1/100 in TBS supplemented with 10% rabbit serum (Chemicon, Heule, Belgium) during 1 hour at room temperature.
  • Anti-RA.21 antiserum was diluted 1/50 in TBS and incubated overnight at room temperature, followed by 1 hour incubation at room temperature with HRP-conjugated rabbit anti-human IgG antibody (Dako) diluted 1/80 in TBS. Staining was performed with DAB substrate.
  • TBS and TBS supplemented with 0.05%TritonX-100 were used for in between washing steps. Counterstaining was performed with Gill's haematoxylin (Klinipath). Control staining for each antibody was performed by omitting the primary antibody.
  • Synthetic UH-RA.21 peptide was coupled to AminoLink columns of beaded agarose (MicroLink Protein Coupling Kit, Fisher Scientific) according to the manufacturer's instructions.
  • 100 ⁇ g UH-RA.21 synthetic peptide was covalently coupled to AminoLink Plus Coupling Gel Spin Column by incubation in coupling buffer (0.1 M sodium phosphate, 0.15 NaCI, pH 7.2) with addition of sodium cyanoborohydride solution (5M, in 0.01 M NaOH) during 4 hours at room temperature.
  • the UH-RA.21 coupled column was incubated 4 times with 250 ⁇ l RA serum containing high antibody levels against UH-RA.21 (according to phage ELISA and peptide ELISA). After washing the column with 0.5M NaCI, the bound antibodies were eluted in three fractions with 100 ⁇ l ImmunoPure elution buffer (pH 2.8). Eluates were immediately neutralised by addition of 5 ⁇ l of 1 M Tris (pH 9.0). Eluates and flow- throughs were evaluated for anti-UH-RA.21 antibody levels by peptide ELISA. The concentration of eluted antibody was determined by spectrophotometry and the BCA protein quantification kit. Tables
  • Table 4 Serological Antigen Selection procedure for the early and RF- ACPA- RA pools: increase in phage output:input titers with increasing selection round
  • Table 5 ELISA screening for the panel of 22 candidate phage cDNA clones
  • HLA-DRB1 HLA class Il DR beta 1 BC024269.1 in coding sequence out of frame
  • ADAMTS12 A disintegrin and metalloproteinase with thrombospondin motifs 12
  • NLSAFYDSELFRMNKFS Minichromosome maintenance protein 2 homolog
  • HDLKRKMILQQF* P49736
  • TGFbeta 1 induced transcript 1 protein (043294) 8/10 (80%) NF-kappa-B essential modulator (IKKAP1)
  • Nell VP Machold KP, Eberl G, Stamm TA, Uffmann M, Smolen JS. Benefit of very early referral and very early therapy with disease-modifying anti-rheumatic drugs in patients with early rheumatoid arthritis. Rheumatology (Oxford) 2004; 43(7):906-914.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Cette invention concerne le diagnostic d’affections auto-immunes, plus spécifiquement le diagnostic d’affections rhumatismales, de l’arthrite auto-immune chronique et plus particulièrement le diagnostic de la polyarthrite rhumatoïde. Un ensemble de biomarqueurs peut être utilisé pour détecter si un sujet est atteint de polyarthrite rhumatoïde. L’invention concerne également des méthodes d’identification de ces biomarqueurs.
PCT/EP2009/053369 2008-03-21 2009-03-23 Biomarqueurs de la polyarthrite rhumatoïde WO2009115612A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP09722261.6A EP2307451B1 (fr) 2008-03-21 2009-03-23 Biomarqueurs de la polyarthrite rhumatoïde
US12/737,281 US9683031B2 (en) 2008-03-21 2009-03-23 Biomarkers for rheumatoid arthritis
ES09722261.6T ES2584438T3 (es) 2008-03-21 2009-03-23 Biomarcadores para artritis reumatoide
US15/498,196 US20180030122A1 (en) 2008-03-21 2017-04-26 Biomarkers for rheumatoid arthritis

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US3848108P 2008-03-21 2008-03-21
US61/038,481 2008-03-21

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/737,281 A-371-Of-International US9683031B2 (en) 2008-03-21 2009-03-23 Biomarkers for rheumatoid arthritis
US15/498,196 Division US20180030122A1 (en) 2008-03-21 2017-04-26 Biomarkers for rheumatoid arthritis

Publications (1)

Publication Number Publication Date
WO2009115612A1 true WO2009115612A1 (fr) 2009-09-24

Family

ID=40637981

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/053369 WO2009115612A1 (fr) 2008-03-21 2009-03-23 Biomarqueurs de la polyarthrite rhumatoïde

Country Status (4)

Country Link
US (2) US9683031B2 (fr)
EP (2) EP2307451B1 (fr)
ES (1) ES2584438T3 (fr)
WO (1) WO2009115612A1 (fr)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130084584A1 (en) * 2010-03-31 2013-04-04 Anna Blom Method to detect tissue degradation leading to inflammation
CN103185800A (zh) * 2011-12-30 2013-07-03 深圳市亚辉龙生物科技有限公司 一种检测抗环瓜氨酸多肽抗体的试剂装置及其方法
WO2015169602A1 (fr) * 2014-05-05 2015-11-12 Noviosmart Procédé de diagnostic sérologique de la polyarthrite rhumatoïde
WO2015189638A3 (fr) * 2014-06-12 2016-02-04 Queen Mary University Of London Anticorps
EP3056904A1 (fr) * 2015-02-13 2016-08-17 F. Hoffmann-La Roche AG Procédé pour l'évaluation de l'arthrite rhumatoïde par la mesure de l'anti-CCP et de l'anti-MCM3
EP3056903A1 (fr) * 2015-02-13 2016-08-17 F. Hoffmann-La Roche AG Procédé d'évaluation de l'arthrite rhumatoïde par la mesure de l'anti-CCP et de l'anti-Casp8
US9683031B2 (en) 2008-03-21 2017-06-20 Universiteit Hasselt Biomarkers for rheumatoid arthritis
CN110286230A (zh) * 2019-06-11 2019-09-27 中国医学科学院北京协和医院 一种acpa阴性的ra诊断标志物及其应用
EP3595710A4 (fr) * 2017-03-13 2021-01-13 Duke University Système de présentation d'antigène et procédés de caractérisation de réponses en anticorps

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9387246B2 (en) 2013-09-03 2016-07-12 L. Douglas Graham Treatment methods for rheumatoid arthritis
JP6188929B2 (ja) 2014-04-22 2017-08-30 国立大学法人東北大学 肺高血圧症の検査方法
GB201712071D0 (en) * 2017-07-27 2017-09-13 Nordic Bioscience As Collagen type X alpha-1 assay

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001057182A2 (fr) * 2000-01-31 2001-08-09 Human Genome Sciences, Inc. Acides nucleiques, proteines et anticorps
WO2005064307A2 (fr) * 2003-12-23 2005-07-14 Roche Diagnostics Gmbh Methode de diagnostic de la polyarthrite rhumatoide, consistant a mesurer la concentration d'anti-ccp et d'interleukine 6

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE511116T1 (de) 1991-04-26 1993-04-29 Clonatec S.A., Paris, Fr Antigene, die von polyarthritis rheumatoide antikoerpern erkannt werden, ihre herstellung und ihre anwendungen.
US5585464A (en) 1993-02-19 1996-12-17 Theratech, Inc. Recombinant antigen for diagnosing rheumatoid arthritis
NL1004539C2 (nl) 1996-11-15 1998-05-20 Stichting Tech Wetenschapp Peptide afgeleid van een door auto-antilichamen van patiënten met reumatoïde artritis herkend antigeen, antilichaam daartegen en werkwijze voor het detecteren van auto-immuunantilichamen.
EP1240180A2 (fr) 1999-12-21 2002-09-18 Innogenetics N.V. Peptides permettant de diagnostiquer et de traiter la polyarthrite rhumatoide
GB0107219D0 (en) * 2001-03-22 2001-05-16 Microbiological Res Authority Immunogenic commensal neisseria sequences
DE10347710B4 (de) * 2003-10-14 2006-03-30 Johannes-Gutenberg-Universität Mainz Rekombinante Impfstoffe und deren Verwendung
JP4685369B2 (ja) * 2004-04-30 2011-05-18 独立行政法人科学技術振興機構 関節リウマチ診断用試薬
FI20050814A0 (fi) 2005-08-11 2005-08-11 Procollagen Oy Menetelmä nivelreumassa muodostuvien kollageeniautovasta-aineiden havaitsemiseksi
ES2584438T3 (es) 2008-03-21 2016-09-27 Universiteit Hasselt Biomarcadores para artritis reumatoide

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001057182A2 (fr) * 2000-01-31 2001-08-09 Human Genome Sciences, Inc. Acides nucleiques, proteines et anticorps
WO2005064307A2 (fr) * 2003-12-23 2005-07-14 Roche Diagnostics Gmbh Methode de diagnostic de la polyarthrite rhumatoide, consistant a mesurer la concentration d'anti-ccp et d'interleukine 6

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LEE D M ET AL: "Clinical utility of the anti-CCP assay in patients with rheumatic diseases", INTERNATIONAL JOURNAL OF GYNAECOLOGY & OBSTETRICS, LIMERICK, IR, vol. 62, no. 9, 1 September 2003 (2003-09-01), pages 870 - 874, XP002334951 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9683031B2 (en) 2008-03-21 2017-06-20 Universiteit Hasselt Biomarkers for rheumatoid arthritis
US20130084584A1 (en) * 2010-03-31 2013-04-04 Anna Blom Method to detect tissue degradation leading to inflammation
CN103185800A (zh) * 2011-12-30 2013-07-03 深圳市亚辉龙生物科技有限公司 一种检测抗环瓜氨酸多肽抗体的试剂装置及其方法
CN103185800B (zh) * 2011-12-30 2015-04-08 深圳市亚辉龙生物科技有限公司 一种检测抗环瓜氨酸多肽抗体的试剂装置及其方法
WO2015169602A1 (fr) * 2014-05-05 2015-11-12 Noviosmart Procédé de diagnostic sérologique de la polyarthrite rhumatoïde
CN106459198B (zh) * 2014-05-05 2021-07-20 诺维奥Th私人有限公司 类风湿性关节炎的血清学诊断方法
US10684283B2 (en) 2014-05-05 2020-06-16 Novio Th B.V. Method for the serological diagnosis of rheumatoid arthritis
CN106459198A (zh) * 2014-05-05 2017-02-22 诺维奥斯玛特 类风湿性关节炎的血清学诊断方法
EP3155012A2 (fr) * 2014-06-12 2017-04-19 Queen Mary University of London Autoanticorps associés à la polyarthrite rhumatoïde
US20170129948A1 (en) * 2014-06-12 2017-05-11 Queen Mary University Of London Antibody
US10590188B2 (en) * 2014-06-12 2020-03-17 Queen Mary University Of London Antibody
WO2015189638A3 (fr) * 2014-06-12 2016-02-04 Queen Mary University Of London Anticorps
EP3056903A1 (fr) * 2015-02-13 2016-08-17 F. Hoffmann-La Roche AG Procédé d'évaluation de l'arthrite rhumatoïde par la mesure de l'anti-CCP et de l'anti-Casp8
EP3056904A1 (fr) * 2015-02-13 2016-08-17 F. Hoffmann-La Roche AG Procédé pour l'évaluation de l'arthrite rhumatoïde par la mesure de l'anti-CCP et de l'anti-MCM3
EP3595710A4 (fr) * 2017-03-13 2021-01-13 Duke University Système de présentation d'antigène et procédés de caractérisation de réponses en anticorps
CN110286230A (zh) * 2019-06-11 2019-09-27 中国医学科学院北京协和医院 一种acpa阴性的ra诊断标志物及其应用

Also Published As

Publication number Publication date
US20110212470A1 (en) 2011-09-01
ES2584438T3 (es) 2016-09-27
EP2307451B1 (fr) 2016-05-25
US20180030122A1 (en) 2018-02-01
EP2307451A1 (fr) 2011-04-13
EP3112379A1 (fr) 2017-01-04
US9683031B2 (en) 2017-06-20

Similar Documents

Publication Publication Date Title
US20180030122A1 (en) Biomarkers for rheumatoid arthritis
US11199549B2 (en) MEl'hods and means for diagnosing spondylarthritis using autoantibody markers
Somers et al. Novel autoantibody markers for early and seronegative rheumatoid arthritis
Cavazzana et al. Systemic sclerosis-specific antibodies: novel and classical biomarkers
Amlani et al. Anti-NT5c1A autoantibodies as biomarkers in inclusion body myositis
US11913950B2 (en) Antigens derived from citrullinated 14-3-3 and uses thereof in the diagnosis of rheumatoid arthritis
JP5998318B2 (ja) 新規血管炎の検査方法および検査用試薬
US10746735B2 (en) Marker sequences for diagnosing and stratifying SLE patients
JP5191544B2 (ja) 皮膚筋炎の検出方法および診断キット
US20220365082A1 (en) Method and means for diagnosis of spondyloarthritis
KR20170093141A (ko) 데옥시하이푸신·신타제 유전자를 지표로서 사용하는 동맥 경화 및 암의 검출 방법
JP2014162772A (ja) 抗シトルリン化タンパクヒトIgG抗体およびその用途
Cleutjens et al. Noninvasive diagnosis of ruptured peripheral atherosclerotic lesions and myocardial infarction by antibody profiling
US20110045509A1 (en) Method For Determining The Type of an Inflammatory-Rheumatic Disease in Synovial Fluid
US11779643B2 (en) Methods and compositions for the treatment of an inflammatory bowel disease
WO2023275235A1 (fr) Procédé et moyens de diagnostic de spondylarthrite
Mor-Vaknin et al. High levels of DEK autoantibodies in sera of polyarticular JIA patients and in early flare following cessation of anti-TNF therapy
GB2622246A (en) Antibody assay
Mahler et al. Research article Clinical and serological evaluation of a novel CENP-A peptide based ELISA

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09722261

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009722261

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12737281

Country of ref document: US